Systematic design of drug delivery therapies

https://doi.org/10.1016/j.compchemeng.2007.06.014Get rights and content

Abstract

This paper presents an engineering approach for optimal drug delivery to the human brain. The hierarchical design procedure addresses three major challenges: (i) physiologically consistent geometric models of the brain anatomy, (ii) discovery of unknown transport and metabolic reaction rates of therapeutic drugs by problem inversion, and (iii) a rigorous method for determining optimal parameters for delivering therapeutic agents to desired target anatomy in the brain. The proposed interdisciplinary approach integrates medical imaging and diagnosis with systems biology and engineering optimization in order to better quantify transport and reaction phenomena in the brain in vivo. It will enhance the knowledge gained from clinical data by combining advanced imaging techniques with large scale optimization of distributed systems. The new procedure will allow physicians and scientists to design and optimize invasive drug delivery techniques systematically based on in vivo drug distribution data and rigorous first principles models.

Introduction

An increasing number of people are affected by neurodegenerative diseases of the central nervous system (CNS) such as Parkinson's, Alzheimer's and Huntington's disease (NIH, 2007). Development of more efficient therapies for diseases of the CNS is hampered by difficulties related to administering therapeutic agents to the affected areas deep inside the brain. Many of the larger proteins with promising pharmacological potency in vitro cannot pass the blood brain barrier (BBB), thus never reaching the cells they are targeted to remedy. Direct injection of the drugs via catheters into the region of interest in the brain can effectively by-pass the BBB, and the penetration depths can be enhanced by convection (Morrison, Laske, Bobo, Oldfield, & Dedrick, 1994). This technique imposes a convective bulk flow field in the extracellular space of the porous brain tissues, thus carrying the drug deep into the brain.

Molecular transport properties of new drugs are usually studied by animal experiments. The interpretation of three-dimensional drug distribution data inside the tissues of rats, rabbits or sheep is not amenable to simplistic lumped or one-dimensional mathematical models. Moreover, the enormous size differences between the human and animal brains make the scaling of drug delivery modalities a non-trivial task. Therefore, there exists a need to create a systematic data-driven approach for the effective design of invasive drug delivery therapies that takes into account the geometric, physiological and biochemical complexity of brain–drug interactions.

Novel analytical imaging techniques such as MRI, functional MRI (fMRI), diffusion tensor imaging (DTI), computer tomography (CT) and positron emission tomography (PET) have improved medical diagnosis. However, the existing imaging technologies are serving physicians only in a qualitative sense. Quantitative information such as transport or metabolic reaction properties are typically not derived from images directly. There appears to exist a gap between high quality of imaging data and their use in quantitative analysis. In order to address the open challenges in accurately predicting drug distribution, its metabolic reaction rates and drug clearance through the blood, a new computational approach integrating clinical imaging data with first principles transport equations is presented. The proposed computer aided design methodology depicted in Fig. 1 aims at overcoming three major challenges. The first issue addresses the accurate reconstruction of the three-dimensional brain geometry. A process to seamlessly integrate imaging data with state-of-the-art geometric image reconstruction tools is presented. The second issue concerns the lack of physical and chemical properties such as diffusivity and metabolic uptake for novel drugs. A computational method for the accurate determination of transport and reaction properties from in vivo medical images with novel mathematical programming techniques called transport and kinetic inversion problem (TKIP) will be introduced. The TKIP will be illustrated via a simplified case study for dopamine kinetics. Finally, drug transport and metabolic rate coefficients will be used to determine optimal parameters for drug administration. These design parameters include: optimal placement and orientation of the injection catheter, catheter dimensions, and the number of drug release openings.

Section 2 describes the approach for the accurate reconstruction of the brain anatomy from images. Section 3 introduces the TKIP approach for the estimation of unknown parameters like diffusivity and kinetic reaction rates. Section 4 describes the finite volume method using generalized curvilinear coordinates for unstructured computational grids. Section 5 quantifies the distribution of a trophic factor and the intravenous injection of L-dopa in different regions of the human brain.

Section snippets

Capturing the complex brain geometry

Quantification of transport processes requires accurate reconstruction of the complex brain anatomy. Brain dimensions and physiological differences from subject to subject justify a patient-specific approach. Fig. 2a shows the brain's inner structure as seen in a histological image (Warner, 2001). Fig. 2b displays the reconstructed two-dimensional computational mesh.

This two-dimensional brain model is composed of volumes and boundary surfaces reconstructed from digital images using image

Discovery of unknown transport properties and metabolic rates

The stages of the computer assisted brain analysis methodology are depicted in Fig. 1. The brain geometry reconstruction was discussed above. The solution of the inversion problem using the finite volume method with the sensitivity equations is described next. We propose a computational method to accurately quantify unknown transport drug properties by interpreting concentration profiles displayed by advanced imaging techniques. Experimental measurements of drug concentration or fluid flow

Finite volume method for unstructured computational meshes

The finite volume method (FVM) aims to discretize partial differential equations (PDE) for species concentration and momentum expressed in Eqs. (2) and (3). In contrast to existing solution methods based on fixed point iterations, e.g. Simple algorithm (Patankar, 1980), we use finite volume discretization for direct and explicit evaluation of the transport equations appearing as equality constraints in the inversion problem. The proposed finite volume method is based on the transport of field

Quantitative analysis and design of drug administration

In this section, two drug administration techniques are discussed to illustrate the computer assisted drug therapy. The first example is the intravenous injection of L-dopa, a precursor to dopamine. The intraparenchymal administration of a trophic factor with a molecular weight of 30,100 kg/kmole is described as a second example for targeted drug delivery to the midbrain.

Conclusions

A systematic approach for the design of drug delivery policies has been introduced. It provides insight in the selection of parameters for drug therapies. The novel method consisted of three steps: (i) accurate reconstruction of the brain geometry, (ii) determination of unknown transport and kinetic properties from experimental data, and (iii) prediction of volumes of distribution. Two drug administration techniques were described, intraparenchymal injection of the nerve growth factor and

Acknowledgments

The authors would like to thank Materialise Inc. for providing a trial research license of Mimics image reconstruction software. The Fluent Inc. is acknowledged for supporting the initial phases of the research with trial Fluent and Gambit licenses. Financial support from the Sussman and Asher Foundation is gratefully acknowledged. Megan Mekarski's contributions during her summer REU project were supported by REU supplementary grants from NSF-EEC 0502272 and NSF-DMI 0328134.

References (26)

  • J.R. Barrio et al.

    Biological imaging and the molecular basis of dopaminergic diseases

    Biochemical Pharmacology

    (1997)
  • V. Bahl et al.

    Modeling of event-driven continuous-discrete processes, lecture notes in computer science, Vol. 2034

    (2001)
  • G. Bohm et al.

    3D adaptive tomography using Delaunay triangles and Voronoi polygons

    Geophysical Prospecting

    (2000)
  • M. Braaten et al.

    A study of recirculating flow computation using body-fitted coordinates: Consistency aspects and mesh skewness

    Numerical Heat Transfer

    (1986)
  • S. Chowdhry et al.

    Automatic structure analysis of large scale differential algebraic systems

  • A.R. Conn et al.

    Trust-region methods

    (2000)
  • A.W. Date

    Introduction to computational fluid dynamics

    (2005)
  • F.A.L. Dullien

    Porous media fluid transport and pore structure

    (1979)
  • J.H. Ferziger et al.

    Computational methods for fluid dynamics

    (2002)
  • C.A.J. Fletcher

    Computational fluid dynamics, Vols. 1 and 2

    (1991)
  • A. Gjedde et al.

    Dopa decarboxylase activity of the living human brain

    Proceeding of the National Academy of Sciences USA

    (1991)
  • A.A. Linninger et al.

    Drug delivery into the human brain

  • A.A. Linninger et al.

    Cerebrospinal fluid flow in the normal and hydrocephalic human brain

    IEEE Transactions on Biomedical Engineering

    (2007)
  • Cited by (36)

    • Numerical simulation of magnetic nanoparticle-based drug delivery in presence of atherosclerotic plaques and under the effects of magnetic field

      2020, Powder Technology
      Citation Excerpt :

      The Lagrangian approach was utilized for particle trajectory in the steady-state blood flow. Somayaji et al. [19] adopted an engineering approach to optimize the process of brain drug delivery. MRI images were used to construct a realistic geometry and a numerical-based package for modeling the blood in the vein was used.

    • Backflow-free catheters for efficient and safe convection-enhanced delivery of therapeutics

      2017, Medical Engineering and Physics
      Citation Excerpt :

      As a result of introducing our backflow-free catheters, there will be increased drug concentration at the target area due to greatly reduced extraparenchymal leakage into cerebrospinal fluid-filled spaces. Also, drug distribution geometries inside porous brain tissue following CED will be more predictable, and methods such as those described in our previous papers [29,35–37] will serve to aid in these predictions. Through implementation of our three backflow-free catheter designs, physicians will be able to target specific regions of the brain with improved accuracy, increased drug concentration, and larger drug distribution geometries.

    • On the appropriateness of modelling brain parenchyma as a biphasic continuum

      2016, Journal of the Mechanical Behavior of Biomedical Materials
      Citation Excerpt :

      The most widely-used models either treat the brain parenchyma as a single phase or a biphasic continuum. For example, biphasic theory (originally developed to model the behaviour of soils, and often termed soil consolidation theory) has been used to model the development of hydrocephalus (Kaczmarek et al., 1997; Momjian and Bichsel, 2008; Nagashima et al., 1987; Peña et al., 1999; Smillie et al., 2005; Taylor and Miller, 2004; Sobey and Wirth, 2006, Wirth and Sobey, 2006, 2009; Cheng and Bilston, 2010) and brain deformations during neurosurgery (Lunn et al., 2006; Miga et al., 1999, 2000; Paulsen et al., 1999; Platenik et al., 2002) as well as to understand phenomena such as convection enhanced drug delivery (Ding et al., 2010; Sampson, 2009; Vogelbaum et al., 2007; Sampson et al., 2007a, 2007b, 2007c; Song and Lonser, 2008; Jagannathan et al., 2008; Szerlip et al., 2007; Morrison et al., 2007; Lonser et al., 2007b, 2007a; Murad et al., 2007, 2002; Croteau et al., 2005; Degen et al., 2003; Sarntinoranont et al., 2003b, 2006, 2003a, 2003c; Raghavan, 2010; Linninger et al., 2008b, 2008a; Somayaji et al., 2008; Astary et al., 2010; Chen et al., 2008, 2007; Chen and Sarntinoranont, 2007). Single-phase continuum theory has been used to model brain injury (King, 2000; Yang and King, 2003; Zhang et al., 2001b, 2001a, 2002, 2004; Donnelly and Medige, 1997; Bilston et al., 2001; Brands et al., 2004; Hrapko et al., 2006, 2009; Nicolle et al., 2004; Ning et al., 2006; Shen et al., 2006; Takhounts et al., 2003), and more recently has been applied to the analysis of hydrocephalus (Dutta-Roy et al., 2008), modelling neurosurgery (Wittek et al., 2007) and surgical simulation (e.g. needle insertion) (Miller et al., 2010).

    View all citing articles on Scopus
    View full text